Quantification of F -isoprostanes as a biomarker of oxidative stress 2

advertisement
PROTOCOL
Quantification of F2-isoprostanes as a biomarker of
oxidative stress
Ginger L Milne, Stephanie C Sanchez, Erik S Musiek & Jason D Morrow
Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, 37232-6602, USA. Correspondence should be addressed to J.D.M.
(jason.morrow@vanderbilt.edu).
Published online 8 February 2007; doi:10.1038/nprot.2006.375
Oxidant stress has been implicated in a wide variety of disease processes. One method to quantify oxidative injury is to measure
lipid peroxidation. Quantification of a group of prostaglandin F2α-like compounds derived from the nonezymatic oxidation of
arachidonic acid, termed the F2-isoprostanes (F2-IsoPs), provides an accurate assessment of oxidative stress both in vitro and
in vivo. In fact, in a recent independent study sponsored by the National Institutes of Health (NIH), F2-IsoPs were shown to be
the most reliable index of in vivo oxidant stress when compared against other well known biomarkers. This protocol details our
laboratory’s method to quantify F2-IsoPs in biological fluids and tissues using gas chromatography-mass spectrometry (GC-MS).
This procedure can be completed for 12–15 samples in 6–8 h.
INTRODUCTION
Free radicals, largely derived from molecular oxygen, have been confounded by the potential contribution of local IsoP production
implicated in a variety of human conditions and diseases, includ- in the kidney, although the extent to which this occurs is unclear. In
ing atherosclerosis and associated risk factors, cancer, neurodegen- light of this issue, we have identified the primary urinary metaboerative diseases and aging. Damage to tissue biomolecules by free lite of 15-F2t-IsoP to be 2,3-dinor-5,6-dihydro-15-F2t-IsoP, and we
radicals is postulated to be a major contributor to the pathophysi- have developed a highly sensitive and accurate mass spectrometric
ology of oxidative stress1,2. Measuring oxidative stress in humans assay to quantify this molecule6. Thus, the quantification of 2,3requires accurate quantification of either free radicals or damaged dinor-5,6-dihydro-15-F2t-IsoP might represent a truly noninvabiomolecules. The targets of free radical–mediated oxidant injury
sive, time-integrated measurement of systemic oxidation status
include lipids, proteins and DNA. A number of methods exist to that can be applied to living subjects.)
quantify free radicals and their oxidation products, although many
Normal levels of F2-IsoPs in healthy humans have been defined
of these techniques suffer from lack of sensitivity and specific- (Table 1)5,7,8. Defining these levels is particularly important
ity, especially when used to assess oxidant stress status in vivo. In in that it allows for an assessment of the effects of diseases on
the Biomarkers of Oxidative Stress Study (BOSS), a recent multi- endogenous oxidant tone and permits the determination of the
investigator study sponsored by the National Institutes of Health extent to which various therapeutic interventions affect levels
(NIH), it was found that the most accurate method to assess of oxidant stress. Elevations of F2-IsoPs in human body fluids
in vivo oxidant stress status is the quantification of plasma or uri- and tissues have been found in a diverse array of human disornary F2-isoprostanes (F2-IsoPs)3. First discovered by our labora- ders, some of which include atherosclerosis, hypercholesteroltory in 1990 (ref. 4), F2-IsoPs, also referred to as 8-iso-PGF2α, are emia, diabetes, obesity, cigarette smoking, neurodegenerative
a series of prostaglandin F2α-like compounds produced by the free diseases, rheumatoid arthritis and many others. Furthermore,
radical–catalyzed peroxidation of arachidonic acid independent of
treatments for some of these conditions, including antioxithe cyclooxygenase. Figure 1 shows the structures of arachidonic dant supplementation, antidiabetic treatments, cessation of
acid and these oxidation products.
smoking and even weight loss, have been shown to decrease
F2-IsoPs are stable, robust molecules detectable in all human tis- production of F2-IsoPs. Thus, the clinical utility of F2-IsoPs has
sues and biological fluids analyzed, including plasma, urine, bron- been great and continues to grow in importance.
choalveolar lavage fluid, cerebrospinal fluid
and bile5. The quantification of F2-IsoPs in
urine and plasma, however, is most convenient and least invasive. And, based on
available data, quantification of these compounds in either plasma or urine is representative of their endogenous production
and thus gives a highly precise and accurate index of in vivo oxidant stress. Analysis
of these compounds in urine in particular
is an index of systemic or ‘whole body’ oxidant stress integrated over time. (The mea- Figure 1 | Structures of arachidonic acid and of its oxidation products, 5-series F -IsoPs, 12-series
2
surement of free F2-IsoPs in urine can be F2-IsoPs, 8-series F2-IsoPs, and the analyte measured in this protocol, 15-series F2-IsoPs.
NATURE PROTOCOLS | VOL.2 NO.1 | 2007 | 221
PROTOCOL
Table 1 | Basal levels of free F2-IsoPs in various body fluids and
tissues from normal humans.
Body fluid
Level (mean ± 1 s.d.)
Plasma
35 ± 6 pg ml–1
Urine
1.6 ± 0.6 ng mg–1 creatinine
Cerebrospinal fluid
23 ± 1 pg ml–1
Several methods have been developed to quantify the F2-IsoPs.
Our laboratory uses a gas chromatography–negative ion chemical
ionization–mass spectrometry (GC-NICI-MS) approach employing stable isotope dilution that will be detailed herein5. For quantification purposes, we measure the F2-IsoP, 15-F2t-IsoP and other
F2-IsoPs that co-elute with this compound. The advantages of this
technique over other approaches include its high sensitivity and
specificity, which yields quantitative results in the low picogram
range. Its drawbacks are that it is labor intensive and requires considerable expenditures on equipment.
Note that different investigators, including FitzGerald and colleagues, have developed several alternative mass spectrometric
assays9–11. Like our assay, these methods quantify F2-IsoPs using
stable isotope dilution GC-NICI-MS and require solid-phase
extraction using a C18 column, thin-layer chromatography (TLC)
purification and chemical derivatization. These assays, however,
measure F2-IsoP isomers other than 15-F2t-IsoP but are comparable to ours in terms of utility. In addition to these gas chromatography–mass spectrometry (GC-MS) assays, a number of liquid chromatography–mass spectrometry (LC-MS) methods for
F2-IsoPs have been developed. One advantage of LC-MS methods
is that the sample preparation for analysis is simpler than that for
GC-MS because it requires no derivatization of the molecule. The
method reported earlier this year by Taylor and colleagues is the
first of these LC-MS methods to be validated for quantitation of
IsoPs in biological fluids12. A concern with LC-MS assays, however,
MATERIALS
REAGENTS
• [2H4]-15-F2t-IsoP (8-iso-PGF2α) internal standard (Cayman Chemical, cat. no.
316351)
•15-F2t-IsoP (8-iso-PGF2α) (Cayman Chemical, cat. no. 16350)
• Butylated hydroxytoluene (BHT) (Sigma-Aldrich, cat. no. B1378)
• Pentafluorobenzyl bromide (PFBB) (Sigma-Aldrich, cat. no. 10105-2)
• N,N′-Diisopropylethylamine (DIPE) (Sigma-Aldrich, cat. no. D3887)
• N,O-bis(trimethylsilyl)trifluoroacetamide (BSTFA) (Supelco, cat. no. 33084)
• Dimethyformamide (DMF) (Sigma-Aldrich, cat. no. 6407) (store over calcium
hydride to prevent water accumulation)
• Undecane (Sigma-Aldrich) (store over calcium hydride to prevent water
accumulation)
• Ultrapure water (triply distilled or its equivalent)
• Methanol
• Chloroform (containing ethanol as a preservative)
• Ethyl acetate
• Heptane
• Acetonitrile
• Ethanol
• HCl (American Chemical Society certified or equivalent grade)
• Sodium chloride
222 | VOL.2 NO.1 | 2007 | NATURE PROTOCOLS
relates to the limits of detection in biological fluids, which are often
higher than those employing GC-MS.
Alternative methods also have been developed to quantify
IsoPs using immunological approaches5,13. Antibodies have been
generated against 15-F2t-IsoP, and at least three immunoassay
kits are available commercially. A potential drawback of these
methods is that information regarding their precision and accuracy is currently limited. In addition, little data exist comparing IsoP levels determined by immunoassay to MS. Analogous
to immunological methods to quantify cyclooxygenase-derived
PGs, it might be predicted that immunoassays for IsoPs will suffer from a lack of specificity. Furthermore, the sensitivity and/or
specificity of these kits might vary substantially between manufacturers. Although mass spectrometric methods of IsoP quantification are considered the best methods for analysis, immunoassays have expanded research in this area due to their low cost
and relative ease of use.
Experimental design
As can be noted from this protocol, quantifying F2-IsoPs using this
methodology is complicated. Further, arachidonic acid in biological samples is susceptible to ex vivo oxidation, which can generate
F2-IsoPs. In the processing of plasma samples and tissues samples,
which both contain an abundance of arachidonic acid, take care
when the samples are obtained and during processing to prevent
ex vivo oxidation. For best results, samples should be flash frozen
in liquid nitrogen immediately upon collection and not thawed
until analysis. For those interested in setting up this assay in their
lab, investigators should first undertake studies with unoxidized
control samples to ensure that F2-IsoPs are not generated during
sample processing. Also, each investigator should determine assay
precision and accuracy before beginning routine sample analysis.
Finally, note that experienced and trained personnel are required
to set up and operate the GC-MS.
• Potassium hydroxide pellets
• Anhydrous sodium sulfate
• 10% Phosphomolybdic acid in ethanol (Sigma-Aldrich, cat. no. P4869)
EQUIPMENT
• React-a-vials with Teflon-lined cap (Supelco, cat. no. 33299)
• Blade homogenizer-PTA 10S generator (Brinkman)
• Nitrogen gas
• C-18 plus solid-phase extraction (or Sep-Pak) cartridges; each cartridge
contains 500 mg of C-18 (Waters, cat. no. WAT036575)
• Silica plus solid-phase extraction (or Sep-Pak) cartridges; each cartridge
contains 500 mg of silica (Waters, cat. no. WAT036580)
• Disposable plastic syringes (10 ml) (Laboratory Supply, cat. no. SMJ512878)
• Glass scintillation vials (20 ml)
• TLC plates: LK6D silica, marked lanes, glass backed (Whatman, cat. no.
WC486562IV)
• Dessicator
• Glass TLC tank (11.5 in w × 10 in h × 3.5 in d)
• TLC paper, cut to 11 in w × 9 in h (VWR, cat. no. 28298-020)
• Capillary GC column (DB-1701, Agilent, cat. no. 21512067)
• GC-MS with capabilities for NICI-MS
REAGENT SETUP
pH 3 water (0.002N HCl) Mix 40 ml 1N hydrochloric acid with 20 L of water.
PROTOCOL
Folch solution Combine 2 volumes of chloroform with 1 volume of
methanol. Dissolve BHT crystals in solution to make a final concentration of
0.005% BHT (wt/vol). Cool to 4 °C and store solution in the dark in a brown
bottle to prevent light degradation.
PFBB solution Dilute PFBB to 10% (vol/vol) in dry acetonitrile. Store over
calcium hydride to keep solution free of water. ! CAUTION PFBB is a potent
lachrymator. Do not work outside of a well-ventilated fume hood.
DIPE solution Dilute DIPE to 10% (vol/vol) in acetonitrile. Store over
calcium hydride to keep solution free of water.
Internal standard The internal standard [2H4]-15-F2t-IsoP is quantified
using a 5-point calibration curve. Each sample contains 0.50 ng of 15-F2t-IsoP,
which has been accurately quantified by weighing, and either 0.05 ng, 0.25 ng,
0.50 ng, 2.50 ng or 5.00 ng of [2H4]-15-F2t-IsoP. Actual [2H4]-15-F2t-IsoP/15F2t-IsoP ratios are compared with the expected ratios to quantify the [2H4]15-F2t-IsoP.
TLC standard, 15-F2t-IsoP methyl ester Dissolve 1 mg of 15-F2t-IsoP
(fatty acid) in 50 ml of dry methanol (solution will be clear). Methylate by
adding dry diazomethane dropwise into the solution until the solution is a
permanent yellow color. Allow to stand 5 min. Dry solution under a stream
of nitrogen. For use, dilute to a concentration 1 mg ml–1 in 3/2 chloroform/
methanol (vol/vol).
EQUIPMENT SETUP
TLC plate preparation Prewash all TLC plates with a solution of ethyl
acetate and ethanol (90:10, vol/vol). One hour before using plates, dry
in a 90 °C oven for 10 min and then cool in a dessicator before sample
application.
GC-MS setup For quantification of F2-IsoPs by GC-MS, we routinely use
an Agilent 5973 mass spectrometer with a computer interface, but other
mass spectrometers can be utilized. The F2-IsoPs are chromatographed on
a 15-meter DB1701 fused silica capillary column because we have found
this column gives excellent separation of individual regioisomers compared
to other columns. The column temperature is programmed from 190 °C
to 300 °C at 20 °C per mm. Methane is used as the reagent gas, and helium
is used as the carrier gas for NICI. The ion source temperature is 200 °C.
The ion monitored for endogenous F2-IsoPs is the carboxylate anion m/z
569 (M-181, loss of CH2C6F5). The corresponding carboxylate anion for
the deuterated intemal standard is m/z 573. Each day the sensitivity of the
mass spectrometer is checked by injecting a standard consisting of 40 pg
each PGF2α and [2H4]-15-F2t-IsoP. Note that PGF2α elutes at a sufficiently
different retention time from the F2-IsoPs quantified using this procedure.
Therefore, this cyclooxygenase-derived prostaglandin does not interfere with
the signal of the non-cyclooxygenase-derived IsoPs.
PROCEDURE
Sample preparation
1| F2-IsoPs can be quantified in biological samples from a variety of sources to afford an accurate index of in vivo oxidant
stress status. In plasma, the present methodology measures only free fatty acid. Although other methodologies can quantify
esterified F2-IsoPs, with values obtained for free F2-IsoPs added to that for esterified F2-IsoPs to measure total F2-IsoPs
in plasma, we have found no advantage in measuring total rather than free F2-IsoPs. Measuring F2-IsoPs in a 24-h urine
collection also represents a reliable probe of the oxidant stress status. With regard to tissue samples, given that F2-IsoPs
from biological sources can only be quantified as free compounds using GC-MS (Morrow et al.7), to measure levels of F2-IsoPs
esterified in glycerophospholipids in tissues, the glycerophospholipids must first be extracted from the sample and then be
subjected to alkaline hydrolysis to release free F2-IsoPs as described below in option C. We recommend preparing blood, urine
and tissue samples for F2-IsoPs quantitation by following the steps in options A, B and C, respectively.
(A) Plasma
(i) Collect blood (5–10 ml) in a tube containing EDTA. (An example of an appropriate collection tube is cat. no. BD367844
from Laboratory Supply Company. This is a 4 ml tube containing 7.2 mg of EDTA.)
(ii) Centrifuge 10 min at 4,000g to yield plasma. 2–3 ml of plasma is necessary for the quantification of F2-IsoPs.
■ PAUSE POINT Store samples at –80 °C until analysis.
(iii) For analysis, after thawing, dilute plasma in 7 ml of pH 3 water and acidify to pH 3 with 1N HCl.
▲ CRITICAL STEP As plasma contains a large amount of arachidonic acid, which can oxidize ex vivo to generate F2-IsoPs,
the samples should be frozen at –80 °C immediately. It is imperative that samples not be thawed before analysis.
(B) Urine
(i) Volunteers should collect all urine voided during a 24-h period in a sterile container, storing it at 4 °C between
collections.
■ PAUSE POINT Store sample at –80 °C until analysis.
(ii) For analysis, after thawing, dilute 0.250 ml urine in 10 ml pH 3 water and acidify to pH 3 with 1N HCl.
(C) Tissue samples
(i) Weigh out 50–100 mg of either fresh tissue or frozen tissue.
▲ CRITICAL STEP Arachidonic acid in tissue samples can oxidize ex vivo to generate F2-IsoPs. If tissue is not to be used
immediately after collection, flash freeze the sample in liquid nitrogen and store at –80 °C.
(ii) Add tissue to 20 ml of ice-cold Folch solution in a 50 ml centrifuge tube with cap. (If sample is <15 mg, reduce the
volume of Folch solution to 10 ml.)
▲ CRITICAL STEP Polypropylene tubes are recommended because polystyrene is not resistant to chloroform. Keep on
ice.
(iii) Homogenize tissue with blade homogenizer at full speed for 30 s or until fully homogenized.
(iv) Flush centrifuge tube with a stream of nitrogen or argon for 30–60 s to remove air from tube, then cap. Let solution
stand at room temperature (22–25 °C) for 1 h to allow maximal extraction of lipids from ground tissue. Shake tube
occasionally for several seconds during this period of time.
(v) Add 4 ml aqueous NaCl (0.9%) prepared in ultrapure water. Vortex or shake vigorously for 1 min at room temperature
(22–25 °C). (If tissue sample is < 15 mg, reduce volume of NaCl solution to 2 ml.)
NATURE PROTOCOLS | VOL.2 NO.1 | 2007 | 223
PROTOCOL
(vi) Centrifuge for 10 min in a tabletop centrifuge at room temperature (22–25 °C) to separate aqueous and organic
layers. Following centrifugation, a semisolid protein layer should have formed between the upper (aqueous) and lower
(organic) layers.
(vii) After centrifugation, carefully pipette off the top aqueous layer and discard. Carefully remove the lower organic layer
from under the intermediate semisolid protein layer and transfer it to a 50 ml conical tube. Evaporate under nitrogen
stream in an analytical evaporation unit until dry.
(viii) Resuspend lipids in 4 ml methanol containing 0.005% BHT and vortex. Next, add 4 ml aqueous KOH (15%, wt/vol). (If
sample originally weighed <15 mg, lipid extract can be resuspended in 1 ml methanol to which 1 ml KOH is added.)
Vortex, purge flask with nitrogen and cap. Incubate mixture at 37 °C for 20 min.
(ix) After incubation, acidify the mixture to pH 3 with 1N HCl and dilute the mixture to 80 ml with pH 3 water.
▲ CRITICAL STEP It is important to dilute the methanol in this solution to 5% or less to ensure proper extraction of
F2-IsoPs in the subsequent purification procedure. It is also important to continue with the steps under ‘Sample
purification for mass spectrometric analysis’. The sample should not be stored in this form because of the potential
for oxidation.
Sample purification for mass spectrometric analysis
2| To sample accurately add 1 ng of the internal standard, [2H4]-15-F2t-IsoP, and vortex.
▲ CRITICAL STEP To ensure accuracy, always add internal standard with an accurate syringe rather than a pipetteman.
3| Connect one C-18 Sep-Pak cartridge to a 10 ml disposable syringe per sample and precondition each cartridge with
5 ml methanol and 7 ml pH 3 water.
4| Apply acidified lipid mixture to the preconditioned Sep-Pak cartridge.
▲ CRITICAL STEP It is important that samples be corrected to pH 3 or slightly below in order for the carboxylate moiety to
be protonated. If not protonated, the compounds will not chromatograph correctly on the Sep-Pak cartridges.
▲ CRITICAL STEP Care must be taken to avoid loss of IsoPs when applying the sample to the cartridge. The sample should
be pushed through the column at ~1–2 ml per min, such that individual drops emerge from the Sep-Pak. Slightly more
vigorous fluid flow is acceptable during the subsequent wash and elution steps, but a steady stream is not recommended.
5| Wash cartridge, first with 10 ml pH 3 water and then with 10 ml heptane.
6| Elute F2-IsoPs from the cartridge with 10 ml ethyl acetate/heptane (50:50, vol/vol) into a glass scintillation vial.
7| Add 5 g anhydrous sodium sulfate to the vial and swirl gently for 10 s. This step removes residual water from the
eluant. Decant eluant into another scintillation vial away from the sodium sulfate.
8| Remove used C-18 Sep-Pak cartridges from syringes and discard. Connect one silica Sep-Pak cartridge for each syringe
per sample and precondition cartridge with 5 ml ethyl acetate.
9| Apply eluant from C-18 Sep-Pak cartridge to silica Sep-Pak cartridge in the same manner as in Step 4.
10| Wash the cartridge with 5 ml ethyl acetate, then elute F2-IsoPs from the silica Sep-Pak cartridge with 5 ml ethyl
acetate/methanol (1:1, vol/vol) into a react-a-vial.
11| Evaporate eluant under nitrogen in the analytical evaporation unit.
12| Convert F2-IsoPs to the corresponding pentaflourobenzyl esters (Fig. 2). (This derivatization facilitates compound
analysis by GC-MS). Add 40 µL of 10% (vol/vol) PFBB in acetonitrile and 20 µL 10% (vol/vol) DIPE in acetonitrile to the
reactivial, vortex briefly and incubate for 20 min at 37 °C.
! CAUTION PFBB is a potent lachrymator. Do not work outside of a well-ventilated fume hood.
13| Prepare TLC tank by adding 97 ml chloroform, 3 ml ethanol and TLC paper to saturate the tank. Allow the tank to
equilibrate for 30 min.
14| Dry sample thoroughly under nitrogen in an analytical evaporation unit in a fume hood and resuspend material in
50 µL methanol. Vortex briefly.
224 | VOL.2 NO.1 | 2007 | NATURE PROTOCOLS
PROTOCOL
15| Apply mixture from each sample
to a prewashed silica TLC plate. Be
sure to apply only one sample per lane
on each plate. Using a separate TLC
plate, apply ~2–5 µg of the methyl
ester of PGF2α to one lane for use as a
standard.
▲ CRITICAL STEP Avoid applying
sample to the first 1 cm of the plate.
Figure 2 | Derivatization of 15-series F2-isoprostanes for GC-MS analysis.
16| After ensuring that the application solvent has dried, place TLC plates into the TLC tank and chromatograph. When the
solvent front reaches 13 cm on the TLC plate, remove from tank and allow solvent to evaporate. Visualize the TLC standard
by spraying the standard plate with the phosphomolybdic acid solution and then heating on a hot plate. (Do not spray
sample plates.)
17| Scrape silica from the sample TLC plates in the region of the TLC standard (Rf should be ~0.18), scraping from 1 cm
above the middle of the visualized standard to 1 cm below the standard (Fig. 3). Retain plates until after GC-MS analysis.
18| Place scraped silica from each sample into separate microcentrifuge tubes and add 1 ml ethyl acetate to each. Vortex
vigorously for 30 s to extract analytes from the silica, then centrifuge in a benchtop microcentrifuge at 13,000 r.p.m. for
3 min.
19| Carefully remove the ethyl acetate, taking care not to disrupt the silica pellet in the bottom of the tube, and place in a
second microcentrifuge tube.
20| Dry under nitrogen, then add 20 µL BSTFA and 7 µL dry DMF to residue to convert to the trimethylsilyl ether
derivatives (Fig. 2).
21| Vortex well and incubate sample at 37 ºC for 20 min. Dry reagents under nitrogen.
■ PAUSE POINT Samples can also incubate in BSTFA and DMF at room temperature (22–25 °C) overnight.
22| Resuspend sample in 20 µL dry undecane and vortex briefly. Transfer sample
to an autosampler vial for GC-MS analysis.
23| A representative chromatogram obtained from the analysis of F2-IsoPs in
plasma is shown in Figure 4. For quantification purposes, we compare the height
of the peak containing the derivatized 15-F2t-IsoP (m/z 569, peak labeled with
* in Fig. 4) with the height of the deuterated internal standard peak
(m/z 573). Levels of F2-IsoPs in plasma are reported in picograms or nanograms
per milliliter, whereas levels in tissues are reported in nanograms per gram of
tissue. Levels of F2-IsoPs in urine are normalized to creatinine clearance and
reported as nanograms per milligrams of creatinine.
● TIMING
In general, 12–15 samples per day can be processed by an experienced
investigator. Lipid extraction and hydrolysis of this number of tissue samples
requires ~3 h. Sep-Pak purification takes ~1.5 h; drying, derivatization and
TLC purification require ~2 h; and extraction, drying and silylation require 1 h.
Mass spectrometric analysis is automated, and each sample requires ~15 min of
instrument time.
? TROUBLESHOOTING
If the peak signal is extremely low or if no peaks are detected by the mass
spectrometer, the sample should be removed from the autosampler vial, dried
thoroughly under nitrogen and Steps 20–22 should be repeated. If peaks are
still not detected, it is possible that there was a problem with the thin-layer
Solvent
front
PGF2α methyl
ester band
(standard,
Rf ~ 0.18)
2 cm
Area to be
scraped
for F2-IsoPs
Figure 3 | Thin layer chromatography analysis
of the methyl ester of PGF2α, the TLC standard,
visualized with phosphomolybdic acid. Indicated
on the plate are the areas to be scraped to
extract F2-IsoPs.
NATURE PROTOCOLS | VOL.2 NO.1 | 2007 | 225
PROTOCOL
chromatography. Scrape all silica from the bottom half of the
lane, place in microcentrifuge tube and extract with ethyl
acetate as described in Step 17. Continue by repeating Steps
18–22. If no result is obtained, it will be necessary to repeat
the entire analysis with new sample.
If the internal standard is detected at m/z 573 but there
are low or nonexistent peaks at m/z 569, then levels of
F2-IsoPs are below the limit of detection. The limit of
detection for this assay is ~5 pg.
ANTICIPATED RESULTS
Figure 4 shows the selected ion current chromatogram
obtained from the analysis of F2-IsoPs in human plasma.
The peaks in the upper m/z 569 selected ion current
chromatogram represents different endogenous F2-IsoPs. This
pattern of peaks is virtually identical to that obtained from
all other biological fluids and tissues that we have examined
to date. The single peak in the lower m/z 573 chromatogram
represents the [2H4]-15-F2t-IsoP internal standard. For
quantification purposes, the peak denoted by an asterisk
(*), which co-elutes with the internal standard, is routinely
measured. The concentration of F2-IsoPs is calculated using
the ratio of the intensity of this peak to that of the internal
standard. In this sample, the concentration of F2-IsoPs was
calculated to 203 pg ml–1.
This method of the quantification of F2-IsoPs is highly
precise and accurate. The precision is ±6 % and the accuracy
is 96%.
ACKNOWLEDGMENTS Supported by NIH grants DK48831, CA77839, GM154312
and ES13125.
COMPETING INTERESTS STATEMENT The authors declare that they have no
competing financial interests.
Figure 4 | Analysis of F2-IsoPs in human plasma. The m/z 573 ion current
chromatogram represents the [2H4]-15-F2t-IsoP internal standard. The m/z
569 ion current chromatogram represents endogenous F2-IsoPs. The peak in
the upper chromatogram represented by the asterisk (*) is the one routinely
used for quantification of the F2-IsoPs.
6.
7.
Published online at http://www.natureprotocols.com/
Reprints and permissions information is available online at http://npg.nature.
com/reprintsandpermissions
8.
1.
9.
2.
3.
4.
5.
Chisholm, G.M. & Steinberg, D. The oxidative modification hypothesis of
atherogenesis: an overview. Free Radical Biol. Med. 18, 1815–1826 (2000).
Halliwell, B. & Gutteridge, J.M. Role of free radicals and catalytic metal
ions in human disease: an overview. Methods Enzymol. 186, 1–85 (1990).
Kadiiska, M.B. et al. Biomarkers of oxidative stress study II: are oxidation
products of lipids, proteins, and DNA markers of CCl4 poisoning? Free
Radic. Biol. Med. 38, 698–710 (2005).
Morrow, J.D. et al. A series of prostaglandin F2-like compounds are
produced in vivo in humans by a non-cyclooxygenase, free radicalcatalyzed mechanism. Proc. Natl. Acad. Sci. USA. 87, 9383–9387 (1990).
Morrow, J.D. et al. Quantification of the major urinary metabolite of
15-F2t-isoprostane (8-iso-PGF2α) by a stable isotope dilution mass
spectrometric assay. Anal. Biochem. 269, 326–331 (1999).
226 | VOL.2 NO.1 | 2007 | NATURE PROTOCOLS
10.
11.
12.
13.
Morales, C.R., Terry, E.S., Zackert, W.E., Montine, T.J. & Morrow, J.D.
Improved assay for the quantification of the major urinary metabolite
of the isoprostane 15-F2t-isoprostane (8-iso-PGF2α) by a stable isotope
dilution mass spectrometric assay. Clin. Chim. Acta 314, 93–99 (2001).
Morrow, J.D. & Roberts, L.J. 2nd. Mass spectrometric quantification of
F2-isoprostanes in biological fluids and tissues as a measure of oxidant
stress. Meth. Enzymol. 300, 3–12 (1999).
Liang, Y. et al. Quantification of 8-iso-prostaglandin-F2α and 2,3-dinor-8iso-prostaglandin-F2α in human urine using liquid chromatography-tandem
mass spectrometry. Free Radic. Biol. Med. 34, 409–418 (2003).
Pratico, D. et al. IPF2alpha-I: an index of lipid peroxidation in humans.
Proc. Natl. Acad. Sci. USA 95, 3449–3454 (1998).
Rokach, J. et al. Nomenclature of isoprostanes: a proposal. Prostaglandins
54, 853–873 (1997).
Rokach, J. et al. The isoprostanes: a perspective. Prostaglandins 54,
823–851 (1997).
Taylor, A.W., Bruno, R.S., Frei, B. & Traber, M.G. Benefits of prolonged
gradient separation for high-performance liquid chromatography-tandem
mass spectrometry quantitation of plasma total 15-series F-isoprostanes.
Anal. Biochem. 350, 41–51 (2006).
Basu, S. Radioimmunoassay of 8-iso-prostaglandin F2α: an index
for oxidative injury via free radical catalysed lipid peroxidation.
Prostaglandins Leukot. Essent. Fatty Acids 58, 319–325 (1998).
Download